personalised medicine challenges: hype or hope? · source: “personalized medicine: current and...

19
An agency of the European Union Personalised medicine challenges: hype or hope? Presented by Marisa Papaluca

Upload: voxuyen

Post on 04-Aug-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

An agency of the European Union

Personalised medicine challenges: hype or hope?

Presented by Marisa Papaluca

Outline

European Council definition of Personalised medicine and EU Medicines Agencies network

strategy to 2020

EMA: Regulatory science and personalised medicines

European Commission: major initiatives

Few question for discussions

1

2

European Council conclusions on PERSONALISED MEDICINE

European Council DEFINTION OF PERSONALISED MEDICINE

No commonly agreed definition of the term “personalised medicine”.

Widely understood that personalised medicine refers to a:

•medical model using characterisation of individuals' phenotypes and

genotypes (e.g. molecular profiling, medical imaging, lifestyle data) for tailoring

the right therapeutic strategy for the right person at the right time, and/or to

determine the predisposition to disease and/or to deliver timely and targeted

prevention.

• Personalised medicine relates to the broader concept of patient-centred care,

which takes into account that, in general, healthcare systems need to better

respond to patient needs

Joint EU Medicines Agencies network strategy to 2020

• ….Key objectives....

Support patient-focussed innovation and contribute to a vibrant lifescience sector in Europe

4

Source: “Personalized Medicine: Current and Future Perspectives,” Patricia Deverka, MD, Duke University, Institute for Genome

Sciences and Policy; and Rick J. Carlson, JD, University of Washington

Dis

ease

burd

en

Time

Typical current intervention

Earliest clinical detection

Earliest molecular detection

Initiating events

Baseline risk

Decision support tools:

Baseline risk Preclinical progression

Disease initiation and progression

Assess risk Refine assessment

Predict diagnose

Track progression Predict events Inform Therapeutics

Sources of new biomarkers:

Stable genomics: Single nucleotide Polymorphisms Haplotype mapping Gene sequencing

Dynamic genomics: Gene expression

Proteomics Metabolomics Molecular imaging

Therapeutic decision support

Drug

Personalised medicine: direction of travel

5

P T. 2011 July; 36(7): 412-416, 419-422, 450. PMCID: PMC3171815 Pharmacogenomics in Clinical Practice Reality and Expectations, C. Lee Ventola, MS 6

Personalised Medicines: healthcare challenges

Direct spend on cancer care across Europe

€ 26,215

€ 24,646

€ 24,381

€ 23,476

€ 22,887

€ 21,475

€ 21,301

€ 21,036

€ 18,072

€ 17,175

€ 16,478

€ 16,243

€ 15,955

€ 14,623

€ 14,459

€ 13,642

€ 12,526

€ 12,215

€ 11,410

€ 11,314

€ 11,140

€ 10,081

€ 9,891

€ 9,600

€ 9,325

€ 9,177

€ 9,002

€ 8,421

€ 0 € 5,000 € 10,000 € 15,000 € 20,000 € 25,000 € 30,000 € 35,000 € 40,000

Greece

Finland

Luxembourg

Austria

Germany

Poland

Netherlands

Estonia

Ireland

Czech Republic

Slovakia

EU-27

United Kingdom

Spain

France

Malta

Italy

Slovenia

Belgium

Latvia

Romania

Cyprus

Denmark

Bulgaria

Sweden

Hungary

Portugal

Lithuania

Healthcare costs per incident COLORECTAL cancer, adjusted for price diferentials

Primary

Outpatient

A&E

Inpatient

Medicine

Slide by courtesy EAPM

• Promote and protect individuals health:

• identify patients who are most likely to benefit patient selection

• identify patients likely to be at increased risk for sADR (Abacavir)

• identify patient for intensified monitoring e.g. during initiation of treatment

• monitor and adjust treatment (e.g. schedule, dose, discontinuation, DDI)

• Promote Patient-centred sustainable health with targeted treatment, early intervention and prevention: HTA/Payers/PH Authorities promoting and embracing the opportunity?

Personalised medicines utility

8

Figure 2: Number of medicinal products and ratio of medicinal products containing a genomic biomarker (gene) in their product label under “Therapeutic Indication” per year.

The number of pharmacogenomic biomarker in EU product label have been steady between 1999 and 2010 and since then gradually increasing in recent years. Initially, they have been intended for information only, progressing into becoming one of the important determinant for selection of patients likely to benefit from treatment and “more” individualised dose selection. Biomarker information may also be included in the labelling in case of negative selection (i.e., if the biomarker is used to select a population unlikely to respond) or in case of uncertainty about the value of the biomarker but where a negative selection is suspected, e.g. vandetanib.

Outlook: targeted therapies on the increase

9

10

Biomarkers and stratified medicines: more efficient clinical trials

11

Pharmacogenomic information

in drug labels: European

Medicines Agency perspective

The Pharmacogenomics Journal

(2015), 1 – 10

Genomics stratified medicines and clinical trials

• Benefit/risk evaluation and regulatory decision making:

– Retrospective analyses versus BM utility prospective validation/subgroups

• Multiplicity issues

• Handling of missing data

• Studies in BM-negative patients: why and when are they needed?

• Emerging new clinical trials designs:

• Adaptive designs

• Umbrella and Basket trials

• Algorithm based trials

• Possibility of using data derived from several independent studies? Pre-competitive research, Open science and new (BIG) data sources

• HTAs acceptance?

12

Towards Personalised Medicines: regulatory science challenges

13

Oversight of the quality and use of molecular tests in the life-cycle of stratified medicines (Implementation of Guidelines on PG and PK, Good Genomic

Practices, Guidelines on genomic BM and drugs co-development, PG methodology in PhVG ICH E18 genomic samples and data handling, etc.)

Consider methodology implication for drug clinical development of Next Generation Sequencing (NGS) for clinical use

analysis of a panel of genes (short term)

analysis of whole exome or genome (medium term)

Large Unbiased Sequencing (long term)

Personalised Medicines: regulatory science challenges

From single genomic biomarkers (as drug targets)

- to multiple biomarkers (pts profiling)

increased estimation bias and type I error

Presentation title (to edit, click Insert > Header & Footer) 14

All BM+ combinations

BM+ subgroups

M. Posch (2012) EMA workshop on pharmacogenomics: from science to clinical care (acknowledgments: A. Graf)

Personalised Medicines: regulatory science challenges

pembrolizumab (Keytruda) anti-PD1 IgG4 (humanized) MSD/Dako 22C3 mouse tumor cells (stroma?) melanoma ≥ 1% NSCLC: ≥1%/≥50%

nivolumab (Opdivo) anti-PD1 IgG4 (human) BMS/Dako 28-8 rabbit tumor cells ≥1%/≥5%

Comparison of test results possible?

15

Personalised Medicines: regulatory science challenges

Qualitative/quantitative: meaning of PD‐L1 positive?

• variability of antibody used, protocols

• tumor surface staining and/or infiltrating lymphocytes

• threshold for PD‐L1 positivity by IHC

• performance metrics CDx – detection and cut-off limits, sensitivity, specificity, reproducibility

Specimen?

• archival tissue or recent - FFPE or fresh; resection or biopsy; intratumoral heterogeneity?

• time point: before start of therapy? on-treatment 1, 2 3 m

Exclusion of PD‐L1 negative in CTs?

• Preliminary data % benefit? predictive claims (BM-restricted indication?)

• alternatives for indication? design (mono vs. combo)?

Extrapolations across indications/treatment lines?

16

Biomarkers in Personalised Medicines: challenges

Patients’ priorities and active role to interventional trials with personalised

medicines: what has changed with omics, how it is perceived, what are the

needs and the preferences to address the implications for individual and family?

HCPs’ role in primary and secondary care: how to responsibly participate in

clinical research and improve the interface with research communities (to

validate new biomarkers, new pre-clinical and clinical methodologies)?

Patients and HCPs support to the development of Clinical (big) data gathering

tools for early access to personalised medicines, the development of prescription

support tools and the longitudinal profiling of the individuals(both clinical status

and tests for personalised medicines).

Role of P and HCP in the evaluation, with regulators, HTAs, payers, and

stakeholders, of the impact of personalised medicines on PH: how to define at

an early stage the value(s) of personalised medicines?

Is it Personalised Medicine a tool towards a sustainable health care? 17

EMA network and personalised medicines development

Personalised medicine: building a bridge to future

Thanks for your attention